Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Sci ; 182(1): 107-119, 2021 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-33892499

RESUMO

The unique physicochemical properties of carbon nanomaterials and their ever-growing utilization generate a serious concern for occupational risk. Pulmonary exposure to these nanoparticles induces local and systemic inflammation, cardiovascular dysfunction, and even cognitive deficits. Although multiple routes of extrapulmonary toxicity have been proposed, the mechanism for and manner of neurologic effects remain minimally understood. Here, we examine the cerebral spinal fluid (CSF)-derived peptidomic fraction as a reflection of neuropathological alterations induced by pulmonary carbon nanomaterial exposure. Male C57BL/6 mice were exposed to 10 or 40 µg of multiwalled carbon nanotubes (MWCNT) by oropharyngeal aspiration. Serum and CSFs were collected 4 h post exposure. An enriched peptide fraction of both biofluids was analyzed using ion mobility-enabled data-independent mass spectrometry for label-free quantification. MWCNT exposure induced a prominent peptidomic response in the blood and CSF; however, correlation between fluids was limited. Instead, we determined that a MWCNT-induced peptidomic shift occurred specific to the CSF with 292 significant responses found that were not in serum. Identified MWCNT-responsive peptides depicted a mechanism involving aberrant fibrinolysis (fibrinopeptide A), blood-brain barrier permeation (homeobox protein A4), neuroinflammation (transmembrane protein 131L) with reactivity by astrocytes and microglia, and a pro-degradative (signal transducing adapter molecule, phosphoglycerate kinase), antiplastic (AF4/FMR2 family member 1, vacuolar protein sorting-associated protein 18) state with the excitation-inhibition balance shifted to a hyperexcited (microtubule-associated protein 1B) phenotype. Overall, the significant pathologic changes observed were consistent with early neurodegenerative disease and were diagnostically reflected in the CSF peptidome.


Assuntos
Nanotubos de Carbono , Doenças Neurodegenerativas , Animais , Inflamação/induzido quimicamente , Pulmão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanotubos de Carbono/toxicidade
2.
Neuropharmacology ; 176: 108170, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32479813

RESUMO

Nicotine, the primary psychoactive component in tobacco, plays a major role in the initiation and maintenance of tobacco dependence and addiction, a leading cause of preventable death worldwide. An essential need thus exists for more effective pharmacotherapies for nicotine-use cessation. Previous reports suggest that pharmacological and genetic blockade of CB1 receptors attenuate nicotine reinforcement and reward; while exogenous agonists enhanced these abuse-related behaviors. In this study, we utilized complementary genetic and pharmacologic approaches to test the hypothesis that increasing the levels of the endocannabinoid 2-arachindonoylglycerol (2-AG), will enhance nicotine reward by stimulating neuronal CB1 receptors. Contrary to our hypothesis, we found that inhibition of monoacylglycerol lipase (MAGL), the primary catabolic enzyme of 2-AG, attenuates nicotine conditioned place preference (CPP) in mice, through a non-CB1 receptor-mediated mechanism. MAGL inhibition did not alter palatable food reward or Lithium Chloride (LiCl) aversion. In support of our findings, repeated MAGL inhibition did not induce a reduction in CB1 brain receptor levels or hinder function. To explore the potential mechanism of action, we investigated if MAGL inhibition affected other fatty acid levels in our CPP paradigm. Indeed, MAGL inhibition caused a concomitant decrease in arachidonic acid (AA) levels in various brain regions of interest, suggesting an AA cascade-dependent mechanism. This idea is supported by dose-dependent attenuation of nicotine preference by the selective COX-2 inhibitors valdecoxib and LM-4131. Collectively, these findings, along with our reported studies on nicotine withdrawal, suggest that inhibition of MAGL represents a promising new target for the development of pharmacotherapies to treat nicotine dependence.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Monoacilglicerol Lipases/antagonistas & inibidores , Monoacilglicerol Lipases/metabolismo , Nicotina/administração & dosagem , Recompensa , Tabagismo/metabolismo , Animais , Ansiolíticos/farmacologia , Ácidos Araquidônicos/farmacologia , Benzodioxóis/farmacologia , Agonistas de Receptores de Canabinoides/farmacologia , Condicionamento Clássico/fisiologia , Endocanabinoides/farmacologia , Inibidores Enzimáticos/farmacologia , Glicerídeos/farmacologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Piperidinas/farmacologia , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/metabolismo , Tabagismo/tratamento farmacológico , Tabagismo/psicologia
3.
Addict Biol ; 25(1): e12691, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30378732

RESUMO

Cessation from prolonged use of ∆9 -tetrahydrocannabinol (THC), the primary active compound responsible for the cannabimimetic effects of cannabis, results in a mild to moderate withdrawal syndrome in humans and laboratory animals. Whereas manipulations of the endogenous cannabinoid system (eg, cannabinoid receptors and endocannabinoid regulating enzymes) alter nicotine withdrawal, in this study we asked the reciprocal question. Do nicotinic acetylcholine receptors (nAChRs) modulate THC withdrawal? To assess the role of different nAChR subtypes in THC withdrawal, we used transgenic mouse, preclinical pharmacological, and human genetic correlation approaches. Our findings show that selective α3ß4* nAChR antagonist, AuIB, and α3ß4* nAChR partial agonist, AT-1001, dose-dependently attenuated somatic withdrawal signs in THC-dependent mice that were challenged with the cannabinoid-1 receptor antagonist rimonabant. Additionally, THC-dependent α5 and α6 nAChR knockout (KO) mice displayed decreased rimonabant precipitated somatic withdrawal signs compared with their wild-type counterparts. In contrast, ß2 and α7 nAChR KO mice showed no alterations in THC withdrawal signs. Moreover, deletion of ß2 nAChR did not alter the reduced expression of somatic signs by the preferred α6ß4* antagonist, BulA [T5A;P60]. Finally, the human genetic association studies indicated that variations in the genes that code for the α5, α3, ß4, and α6 nAChRs were associated with cannabis disorder phenotypes. Overall, these findings suggest that α3ß4* and α6ß4* nAChR subtypes represent viable targets for the development of medications to counteract THC dependence.


Assuntos
Dronabinol/farmacologia , Abuso de Maconha/fisiopatologia , Receptores Nicotínicos/efeitos dos fármacos , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Antagonistas de Receptores de Canabinoides/administração & dosagem , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rimonabanto/administração & dosagem
4.
Part Fibre Toxicol ; 16(1): 20, 2019 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-31142334

RESUMO

BACKGROUND: The mechanisms driving systemic effects consequent pulmonary nanoparticle exposure remain unclear. Recent work has established the existence of an indirect process by which factors released from the lung into the circulation promote systemic inflammation and cellular dysfunction, particularly on the vasculature. However, the composition of circulating contributing factors and how they are produced remains unknown. Evidence suggests matrix protease involvement; thus, here we used a well-characterized multi-walled carbon nanotube (MWCNT) oropharyngeal aspiration model with known vascular effects to assess the distinct contribution of nanoparticle-induced peptide fragments in driving systemic pathobiology. RESULTS: Data-independent mass spectrometry enabled the unbiased quantitative characterization of 841 significant MWCNT-responses within an enriched peptide fraction, with 567 of these factors demonstrating significant correlation across animal-paired bronchoalveolar lavage and serum biofluids. A database search curated for known matrix protease substrates and predicted signaling motifs enabled identification of 73 MWCNT-responsive peptides, which were significantly associated with an abnormal cardiovascular phenotype, extracellular matrix organization, immune-inflammatory processes, cell receptor signaling, and a MWCNT-altered serum exosome population. Production of a diverse peptidomic response was supported by a wide number of upregulated matrix and lysosomal proteases in the lung after MWCNT exposure. The peptide fraction was then found bioactive, producing endothelial cell inflammation and vascular dysfunction ex vivo akin to that induced with whole serum. Results implicate receptor ligand functionality in driving systemic effects, exemplified by an identified 59-mer thrombospondin fragment, replete with CD36 modulatory motifs, that when synthesized produced an anti-angiogenic response in vitro matching that of the peptide fraction. Other identified peptides point to integrin ligand functionality and more broadly to a diversity of receptor-mediated bioactivity induced by the peptidomic response to nanoparticle exposure. CONCLUSION: The present study demonstrates that pulmonary-sequestered nanoparticles, such as multi-walled carbon nanotubes, acutely upregulate a diverse profile of matrix proteases, and induce a complex peptidomic response across lung and blood compartments. The serum peptide fraction, having cell-surface receptor ligand properties, conveys peripheral bioactivity in promoting endothelial cell inflammation, vasodilatory dysfunction and inhibiting angiogenesis. Results here establish peptide fragments as indirect, non-cytokine mediators and putative biomarkers of systemic health outcomes from nanoparticle exposure.


Assuntos
Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Exposição por Inalação/efeitos adversos , Pulmão/efeitos dos fármacos , Nanotubos de Carbono/toxicidade , Fragmentos de Peptídeos/sangue , Animais , Biomarcadores/sangue , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/metabolismo , Células Endoteliais/imunologia , Endotélio Vascular/imunologia , Endotélio Vascular/fisiopatologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/imunologia , Expressão Gênica/efeitos dos fármacos , Inflamação , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Nanotubos de Carbono/química
5.
J Neurotrauma ; 36(22): 3092-3102, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31122143

RESUMO

Advancing therapeutics for traumatic brain injury (TBI) remains a challenge, necessitating testable targets with interventions appropriately timed to intercede on evolving secondary insults. Neuroproteomics provides a global molecular approach to deduce the complex post-translational processes that underlie secondary events after TBI. Yet method advancement has outpaced approaches to interrogate neuroproteomic complexity, in particular when addressing the well-recognized temporal evolution of TBI pathobiology. Presented is a detailed account of the temporal neuroproteomic response to mild-moderate rat controlled cortical impact within perilesioned somatosensory neocortex across the first two weeks after injury. Further, this investigation assessed use of artificial neural network and functional enrichment analyses to discretize the temporal response across some 2047 significantly impacted proteins. Results were efficiently narrowed onto ion transporters with phenotypic relevance to abnormal GABAergic transmission and a delayed decline amenable to intervention under managed care conditions. The prototypical target potassium/chloride co-transporter 2 (KCC2 or SLC12A5) was investigated further with the KCC2-selective modulator CLP290. Guided by post-translational processing revealed one-day after insult to precede KCC2 protein loss a day after, CLP290 was highly effective at restoring up to 70% of lost KCC2 localization, which was significantly correlated with recovery of sham-level function in assessed somatosensory behavioral tasks. The timing of administration was important, with no significant improvement observed if given earlier, one-hour after insult, or later when KCC2 protein decline begins. Results portend importance for a detailed post-translational characterization when devising TBI treatments, and support the therapeutic promise of KCC2-targeted CLP290 intervention for positive functional recovery after brain injury.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Terapia de Alvo Molecular/métodos , Pró-Fármacos/farmacologia , Simportadores/efeitos dos fármacos , Simportadores/metabolismo , Animais , Masculino , Redes Neurais de Computação , Proteoma , Proteômica , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Tiazolidinas/farmacologia , Cotransportadores de K e Cl-
6.
Behav Brain Res ; 360: 262-269, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30552947

RESUMO

The objective of the present study is to investigate the role of α4, α5, α6 or ß2 nAChR subunits in the antidepressant-like effect of bupropion. Adult male mice were treated with subcutaneous acute doses of bupropion (3 and 10 mg/kg) 30 min before the forced swim test (FST) in α4, α5, α6, or ß2 nAChR subunit knockout (KO) and wild-type (WT) mice. In addition, the effects of ß2* antagonist dihydro-ß-erythroidine (DHßE, 3 mg/kg) on antidepressant-like effects of bupropion in C57BL/6 J mice were assessed. Our results showed that baseline immobility and climbing time did not differ between KO and corresponding WT mice except for ß2 KO. Bupropion significantly decreased immobility time and increased climbing time in the α4, α6 and ß2 nAChR KO mice in comparison to WT littermates, indicating that lack of these nAChR subunits enhanced antidepressant effects of bupropion. On the contrary, the α5 nAChR subunit deletion did not alter the FST behavior in the bupropion-treated mice. Not only in the transgenic mice, bupropion also showed antidepressant-like effects in the WT mice. In addition, DHßE pretreatment before bupropion administration resulted in decreased immobility time and increased climbing time. Taken together, the present study provides evidence on the involvement of α4*, α6*, and ß2* (* indicates possible presence of other subunits) nAChRs in the antidepressant-like effects of bupropion in the FST.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Bupropiona/farmacologia , Receptores Nicotínicos/metabolismo , Natação/psicologia , Análise de Variância , Animais , Di-Hidro-beta-Eritroidina/farmacologia , Relação Dose-Resposta a Droga , Resposta de Imobilidade Tônica/fisiologia , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Desempenho Psicomotor/efeitos dos fármacos , Receptores Nicotínicos/genética
7.
Proteomics ; 18(24): e1800268, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30474317

RESUMO

Secondhand smoke remains a global concern for children's health. Epidemiological studies implicate exposure to secondhand smoke as a major risk factor for behavioral disorders, yet biological causation remains unclear. Model studies have mainly focused on secondhand smoke impacts to prenatal neurodevelopment, yet juvenile exposure represents a separate risk. Using ion mobility-enhanced data-independent mass spectrometry, the effect of juvenile secondhand smoke exposure on the prefrontal cortex, a principal part of the brain involved in behavioral control, is characterized. The produced dataset includes 800 significantly responsive proteins within the juvenile orbital frontal cortex, with 716 showing an increase in abundance. The neuroproteomic response reflects a prominent perturbation within the glutamatergic synaptic system, suggesting aberrant, disorganized excitation as observed underlying psychiatric disorders. Also disclosed are impacts to GABAergic and dopaminergic systems. Overall, the dataset provides a wealth of detail, facilitating further targeted research into the causal mechanisms underlying behavioral disorders associated with juvenile exposure to secondhand smoke and other environmental pollutants. All MS data have been deposited to the ProteomeXchange consortium with identifier PXD011744.


Assuntos
Exposição Ambiental/análise , Lobo Frontal/metabolismo , Proteoma/análise , Poluição por Fumaça de Tabaco/análise , Animais , Animais Recém-Nascidos , Feminino , Lobo Frontal/efeitos dos fármacos , Ratos
8.
Neuropharmacology ; 118: 38-45, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28279662

RESUMO

Chronic tobacco use dramatically increases health burdens and financial costs. Limitations of current smoking cessation therapies indicate the need for improved molecular targets. The main addictive component of tobacco, nicotine, exerts its dependency effects via nicotinic acetylcholine receptors (nAChRs). Activation of the homomeric α7 nAChR reduces nicotine's rewarding properties in conditioned place preference (CPP) test and i.v. self-administration models, but the mechanism underlying these effects is unknown. Recently, the nuclear receptor peroxisome proliferator-activated receptor type-α (PPARα) has been implicated as a downstream signaling target of the α7 nAChR in ventral tegmental area dopamine cells. The present study investigated PPARα as a possible mediator of the effect of α7 nAChR activation in nicotine dependence. Our results demonstrate the PPARα antagonist GW6471 blocks actions of the α7 nAChR agonist PNU282987 on nicotine reward in an unbiased CPP test in male ICR adult mice. These findings suggests that α7 nAChR activation attenuates nicotine CPP in a PPARα-dependent manner. To evaluate PPARα activation in nicotine dependence we used the selective and potent PPARα agonist, WY-14643 and the clinically used PPARα activator, fenofibrate, in nicotine CPP and we observed attenuation of nicotine preference, but fenofibrate was less potent. We also studied PPARα in nicotine dependence by evaluating its activation in nicotine withdrawal. WY-14643 reversed nicotine withdrawal signs whereas fenofibrate had modest efficacy. This suggests that PPARα plays a role in nicotine reward and withdrawal and that further studies are warranted to elucidate its function in mediating the effects of α7 nAChRs in nicotine dependence.


Assuntos
Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , PPAR alfa/metabolismo , Tabagismo/tratamento farmacológico , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Anestésicos Locais/administração & dosagem , Animais , Benzamidas/farmacologia , Compostos Bicíclicos com Pontes/farmacologia , Cocaína/administração & dosagem , Condicionamento Operante/efeitos dos fármacos , Modelos Animais de Doenças , Fenofibrato/farmacologia , Hipolipemiantes/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Oxazóis/farmacologia , PPAR alfa/agonistas , PPAR alfa/antagonistas & inibidores , Pirimidinas/farmacologia , Autoadministração , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Tirosina/análogos & derivados , Tirosina/farmacologia
9.
Proc Natl Acad Sci U S A ; 114(10): E1968-E1976, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28223486

RESUMO

Pulmonary exposure to multiwalled carbon nanotubes (MWCNTs) causes indirect systemic inflammation through unknown pathways. MWCNTs translocate only minimally from the lungs into the systemic circulation, suggesting that extrapulmonary toxicity may be caused indirectly by lung-derived factors entering the circulation. To assess a role for MWCNT-induced circulating factors in driving neuroinflammatory outcomes, mice were acutely exposed to MWCNTs (10 or 40 µg/mouse) via oropharyngeal aspiration. At 4 h after MWCNT exposure, broad disruption of the blood-brain barrier (BBB) was observed across the capillary bed with the small molecule fluorescein, concomitant with reactive astrocytosis. However, pronounced BBB permeation was noted, with frank albumin leakage around larger vessels (>10 µm), overlain by a dose-dependent astroglial scar-like formation and recruitment of phagocytic microglia. As affirmed by elevated inflammatory marker transcription, MWCNT-induced BBB disruption and neuroinflammation were abrogated by pretreatment with the rho kinase inhibitor fasudil. Serum from MWCNT-exposed mice induced expression of adhesion molecules in primary murine cerebrovascular endothelial cells and, in a wound-healing in vitro assay, impaired cell motility and cytokinesis. Serum thrombospondin-1 level was significantly increased after MWCNT exposure, and mice lacking the endogenous receptor CD36 were protected from the neuroinflammatory and BBB permeability effects of MWCNTs. In conclusion, acute pulmonary exposure to MWCNTs causes neuroinflammatory responses that are dependent on the disruption of BBB integrity.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Barreira Hematoencefálica/efeitos dos fármacos , Portadores de Fármacos/efeitos adversos , Encefalite/prevenção & controle , Nanotubos de Carbono/efeitos adversos , Inibidores de Proteínas Quinases/farmacologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Administração por Inalação , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Antígenos CD36/deficiência , Antígenos CD36/genética , Movimento Celular/efeitos dos fármacos , Encefalite/induzido quimicamente , Encefalite/genética , Encefalite/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fluoresceína/farmacocinética , Corantes Fluorescentes/farmacocinética , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Trombospondina 1/genética , Trombospondina 1/metabolismo , Quinases Associadas a rho/genética
10.
J Neurochem ; 138(6): 806-20, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27344019

RESUMO

Nicotinic acetylcholine receptors (nAChRs) affect multiple physiological functions in the brain and their functions are modulated by regulatory proteins of the Lynx family. Here, we report for the first time a direct interaction of the Lynx protein LY6/PLAUR domain-containing 6 (Lypd6) with nAChRs in human brain extracts, identifying Lypd6 as a novel regulator of nAChR function. Using protein cross-linking and affinity purification from human temporal cortical extracts, we demonstrate that Lypd6 is a synaptically enriched membrane-bound protein that binds to multiple nAChR subtypes in the human brain. Additionally, soluble recombinant Lypd6 protein attenuates nicotine-induced hippocampal inward currents in rat brain slices and decreases nicotine-induced extracellular signal-regulated kinase phosphorylation in PC12 cells, suggesting that binding of Lypd6 is sufficient to inhibit nAChR-mediated intracellular signaling. We further show that perinatal nicotine exposure in rats (4 mg/kg/day through minipumps to dams from embryonic day 7 to post-natal day 21) significantly increases Lypd6 protein levels in the hippocampus in adulthood, which did not occur after exposure to nicotine in adulthood only. Our findings suggest that Lypd6 is a versatile inhibitor of cholinergic signaling in the brain, and that Lypd6 is dysregulated by nicotine exposure during early development. Regulatory proteins of the Lynx family modulate the function of nicotinic receptors (nAChRs). We report for the first time that the Lynx protein Lypd6 binds to nAChRs in human brain extracts, and that recombinant Lypd6 decreases nicotine-induced ERK phosphorylation and attenuates nicotine-induced hippocampal inward currents. Our findings suggest that Lypd6 is a versatile inhibitor of cholinergic signaling in the brain.


Assuntos
Antígenos Ly/metabolismo , Receptores Nicotínicos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Animais Recém-Nascidos , Antígenos Ly/genética , Química Encefálica/genética , Proteínas Ligadas por GPI , Humanos , Técnicas In Vitro , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Células PC12 , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/genética , Lobo Temporal/química , Distribuição Tecidual
11.
Alcohol Clin Exp Res ; 40(1): 62-72, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26727524

RESUMO

BACKGROUND: Ethanol (EtOH) and nicotine abuse are 2 leading causes of preventable mortality in the world, but little is known about the pharmacological mechanisms mediating co-abuse. Few studies have examined the interaction of the acute effects of EtOH and nicotine. Here, we examine the effects of nicotine administration on the duration of EtOH-induced loss of righting reflex (LORR) and characterize the nature of their pharmacological interactions in C57BL/6J mice. METHODS: We assessed the effects of EtOH and nicotine and the nature of their interaction in the LORR test using isobolographic analysis after acute injection in C57BL/6J male mice. Next, we examined the importance of receptor efficacy using nicotinic partial agonists varenicline and sazetidine. We evaluated the involvement of major nicotinic acetylcholine receptor (nAChR) subtypes using nicotinic antagonist mecamylamine and nicotinic α4- and α7-knockout mice. The selectivity of nicotine's actions on EtOH-induced LORR was examined by testing nicotine's effects on the hypnotic properties of ketamine and pentobarbital. We also assessed the development of tolerance after repeated nicotine exposure. Last, we assessed whether the effects of nicotine on EtOH-induced LORR extend to hypothermia and EtOH intake in the drinking in the dark (DID) paradigm. RESULTS: We found that acute nicotine injection enhances EtOH's hypnotic effects in a synergistic manner and that receptor efficacy plays an important role in this interaction. Furthermore, tolerance developed to the enhancement of EtOH's hypnotic effects by nicotine after repeated exposure of the drug. α4* and α7 nAChRs seem to play an important role in nicotine-EtOH interaction in the LORR test. In addition, the magnitude of EtOH-induced LORR enhancement by nicotine was more pronounced in C57BL/6J than DBA/2J mice. Furthermore, acute nicotine enhanced ketamine and pentobarbital hypnotic effects in the mouse. Finally, nicotine enhanced EtOH-induced hypothermia but decreased EtOH intake in the DID test. CONCLUSIONS: Our results demonstrate that nicotine synergistically enhances EtOH-induced LORR in the mouse.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Reflexo de Endireitamento/efeitos dos fármacos , Animais , Azetidinas/farmacologia , Interações Medicamentosas , Hipnóticos e Sedativos/farmacologia , Hipotermia , Ketamina/farmacologia , Mecamilamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Antagonistas Nicotínicos/farmacologia , Pentobarbital/farmacologia , Piridinas/farmacologia , Receptores Nicotínicos/genética , Vareniclina/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/genética
12.
Behav Pharmacol ; 27(1): 50-6, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26292186

RESUMO

Several recent studies have indicated the involvement of calcium-dependent mechanisms, in particular the abundant calcium-activated kinase, calcium/calmodulin-dependent kinase II (CaMKII), in behaviors associated with nicotine dependence in mice. Behavioral and biochemical studies have shown that CaMKII is involved in acute and chronic nicotine behaviors and nicotine withdrawal; however, evidence of a role for CaMKII in nicotine reward is lacking. Thus, the goal of the current study was to examine the role of CaMKII in nicotine reward. Using pharmacological and genetic tools, we tested nicotine conditioned place preference (CPP) in C57Bl/6 mice after administration of CaMKII antagonists and in α-CaMKII wild-type (+/+) and heterozygote (±) mice. CaMKII antagonists blocked expression of nicotine CPP, and the preference score was significantly reduced in α-CaMKII ± mice compared with their +/+ counterparts. Further, we assessed CaMKII activity in the ventral tegmental area (VTA), nucleus accumbens (NAc), prefrontal cortex, and hippocampus after nicotine CPP and found significant increases in CaMKII activity in the mouse VTA and NAc that were blocked by CaMKII antagonists. The findings from this study show that CaMKII mediates nicotine reward and suggest that increases in CaMKII activity in the VTA and NAc are relevant to nicotine reward behaviors.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Recompensa , Comportamento Espacial/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Alimentos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
Neuropharmacology ; 102: 236-43, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26639043

RESUMO

Pain encompasses both a sensory as well as an affective dimension and these are differentially processed in the brain and periphery. It is therefore important to develop animal models to reflect the non-reflexive assays in pain. In this study, we compared effects of the mu opioid receptor agonist morphine, the nonsteroidal anti-inflammatory drug ketoprofen and the kappa receptor opioid agonist U50,488H and antagonist JDTic on acetic acid-induced stretching and acetic acid-induced aversion in the condition place aversion (CPA) test in male ICR mice. Intraperitoneal administration of acetic acid (0.32-1%) was equipotent in stimulating stretching and CPA. Ketoprofen, morphine and U50,488H all inhibited the acid-induced stretching. Ketoprofen and morphine also blocked the acid-induced CPA but U50,488H failed to do so. The reversal ability of ketoprofen and morphine on acid-induced CPA is unique to pain-stimulated place aversion since these drugs failed to reduce non-noxious LiCl-induced CPA. Overall, this study characterized and validated a preclinical mouse model of pain-related aversive behavior that can be used to assess genetic and biological mechanisms of pain as well as improving the predictive validity of preclinical studies on candidate analgesics.


Assuntos
(trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Analgésicos Opioides/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Cetoprofeno/farmacologia , Morfina/farmacologia , Dor Visceral/fisiopatologia , Animais , Aprendizagem da Esquiva/fisiologia , Comportamento Animal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Piperidinas/farmacologia , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/agonistas , Tetra-Hidroisoquinolinas/farmacologia
14.
PLoS One ; 10(9): e0137070, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26355604

RESUMO

Although menthol, a common flavoring additive to cigarettes, has been found to impact the addictive properties of nicotine cigarettes in smokers little is known about its pharmacological and molecular actions in the brain. Studies were undertaken to examine whether the systemic administration of menthol would modulate nicotine pharmacokinetics, acute pharmacological effects (antinociception and hypothermia) and withdrawal in male ICR mice. In addition, we examined changes in the brain levels of nicotinic receptors of rodents exposed to nicotine and menthol. Administration of i.p. menthol significantly decreased nicotine's clearance (2-fold decrease) and increased its AUC compared to i.p. vehicle treatment. In addition, menthol pretreatment prolonged the duration of nicotine-induced antinociception and hypothermia (2.5 mg/kg, s.c.) for periods up to 180 min post-nicotine administration. Repeated administration of menthol with nicotine increased the intensity of mecamylamine-precipitated withdrawal signs in mice exposed chronically to nicotine. The potentiation of withdrawal intensity by menthol was accompanied by a significant increase in nicotine plasma levels in these mice. Western blot analyses of α4 and ß2 nAChR subunit expression suggests that chronic menthol impacts the levels and distribution of these nicotinic subunits in various brain regions. In particular, co-administration of menthol and nicotine appears to promote significant increase in ß2 and α4 nAChR subunit expression in the hippocampus, prefrontal cortex and striatum of mice. Surprisingly, chronic injections of menthol alone to mice caused an upregulation of ß2 and α4 nAChR subunit levels in these brain regions. Because the addition of menthol to tobacco products has been suggested to augment their addictive potential, the current findings reveal several new pharmacological molecular adaptations that may contribute to its unique addictive profile.


Assuntos
Mentol/uso terapêutico , Nicotina/farmacologia , Nicotina/farmacocinética , Tabagismo/tratamento farmacológico , Animais , Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Hipotermia Induzida , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Mentol/administração & dosagem , Mentol/farmacologia , Camundongos Endogâmicos ICR , Nicotina/sangue , Nociceptividade/efeitos dos fármacos , Receptores Nicotínicos/metabolismo , Síndrome de Abstinência a Substâncias , Fatores de Tempo , Tabagismo/complicações
15.
Drug Alcohol Depend ; 146: 7-16, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25479915

RESUMO

BACKGROUND: Inhibition of endocannabinoid catabolic enzymes fatty acid amide hydrolase (FAAH) and/or monoacylglycerol lipase (MAGL) reduces somatic morphine withdrawal signs, but its effects on aversive aspects of withdrawal are unknown. The present study investigated whether Δ(9)-tetrahydrocannabinol (THC), the MAGL inhibitor JZL184, the FAAH inhibitor PF-3845, or the dual FAAH/MAGL inhibitor SA-57 would reduce acquisition of morphine withdrawal-induced conditioned place avoidance (CPA) and jumping. METHODS: Mice were implanted with placebo or 75 mg morphine pellets, 48 h later injected with naloxone or saline and placed in the conditioning apparatus, and assessed for CPA at 72 h. Subjects were also observed for jumping behavior following naloxone challenge. RESULTS: Naloxone (0.056 mg/kg) produced robust CPA in morphine-pelleted, but not placebo-pelleted, mice. Morphine pretreatment prevented the occurrence of withdrawal CPA and withdrawal jumping, while clonidine (an α2 adrenergic receptor agonist) only blocked withdrawal CPA. THC, JZL184, and SA-57 significantly reduced the percentage of mice that jumped during the conditioning session, but did not affect acquisition of withdrawal CPA. PF-3845 did not reduce morphine withdrawal CPA or jumping. Finally, neither THC nor the endocannabinoid catabolic enzyme inhibitors in non-dependent mice elicited a conditioned place preference or aversion. CONCLUSIONS: These findings suggest that inhibiting endocannabinoid catabolic enzymes reduces somatic morphine withdrawal signs, but not aversive aspects as inferred in the CPA paradigm. The observation that non-dependent mice administered inhibitors of endocannabinoid degradation did not display place preferences is consistent with the idea that that endocannabinoid catabolic enzymes might be targeted therapeutically, with reduced risk of abuse.


Assuntos
Endocanabinoides/metabolismo , Inibidores Enzimáticos/farmacologia , Morfina/efeitos adversos , Síndrome de Abstinência a Substâncias/psicologia , Acetamidas/farmacologia , Amidoidrolases/antagonistas & inibidores , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Benzodioxóis/farmacologia , Carbamatos/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Dronabinol/farmacologia , Masculino , Camundongos , Monoacilglicerol Lipases/antagonistas & inibidores , Naloxona/farmacologia , Piperidinas/farmacologia , Piridinas/farmacologia
16.
Neuropharmacology ; 85: 67-72, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24859605

RESUMO

Metabolism of nicotine to inactive cotinine by hepatic enzyme CYP2A6 is the principal pathway by which active nicotine is removed from circulation. We therefore hypothesized that inhibition of mouse CYP2A5, the ortolog of human CYP2A6, by methoxsalen (8-methoxypsoralen) alter dependence-related behaviors of nicotine in the mouse. Conditioned place preference (CPP) test was used to assess the appetitive reward-like properties and precipitated nicotine withdrawal to assess physical (somatic and hyperalgesia) and affective (anxiety-related behaviors) measures. The nicotine plasma levels were also measured with or without methoxsalen pretreatment. Methoxsalen (15 and 30 mg/kg, intraperitoneally) pretreatment enhanced nicotine-induced preference in mice (p<0.05). However, there was a lack of enhancement of nicotine in the CPP test after the highest dose of the CYP-2A5 inhibitor. Similarly to the CPP results, repeated administration of methoxsalen increased the intensity of mecamylamine-precipitated withdrawal signs. The potentiation of nicotine preference and withdrawal intensity by methoxsalen was accompanied by significant increase in nicotine plasma levels in mice (p<0.05). Finally, methoxsalen enhanced the ability of a very low dose of nicotine (0.05 mg/kg) to reverse withdrawal signs in mice undergoing spontaneous withdrawal after chronic nicotine infusion (p<0.05). In conclusion, inhibition of nicotine metabolism by methoxsalen alters the behavioral effects of nicotine in the mouse. Combining CYP2A6 inhibitors with low dose nicotine replacement therapies may have a beneficial role in smoking cessation because it will decrease the drug elimination rate and maintain plasma and brain nicotine levels.


Assuntos
Inibidores Enzimáticos/farmacologia , Metoxaleno/farmacologia , Recompensa , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Tabagismo/tratamento farmacológico , Animais , Ansiedade/tratamento farmacológico , Ansiedade/fisiopatologia , Hidrocarboneto de Aril Hidroxilases/antagonistas & inibidores , Hidrocarboneto de Aril Hidroxilases/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Família 2 do Citocromo P450 , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hiperalgesia/tratamento farmacológico , Hiperalgesia/fisiopatologia , Masculino , Mecamilamina/farmacologia , Camundongos Endogâmicos ICR , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Nicotina/sangue , Nicotina/farmacologia , Agonistas Nicotínicos/sangue , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Índice de Gravidade de Doença , Síndrome de Abstinência a Substâncias/fisiopatologia , Tabagismo/fisiopatologia
17.
Psychopharmacology (Berl) ; 229(4): 591-601, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23652588

RESUMO

RATIONALE: Whereas cannabinoid CB1 receptors have long been known to contribute to the rewarding effects and dependence liability of many drugs of abuse, recent studies have implicated the involvement of cannabinoid CB2 receptors. OBJECTIVE: Here, we evaluated the role of CB2 receptors in the rewarding properties of nicotine, as assessed in the conditioned place preference (CPP) paradigm and mecamylamine-precipitated withdrawal in nicotine dependent mice. METHODS: Using complementary pharmacological and genetic approaches, we investigated the involvement of CB2 receptors in nicotine- and cocaine-induced CPP in mice and mecamylamine-precipitated withdrawal in nicotine-dependent mice. We also determined whether deletion of CB2 receptors affects nicotine-induced hypothermia and hypoalgesia. RESULTS: Nicotine-induced (0.5 mg/kg) CPP was completely blocked by selective CB2 antagonist, SR144528 (3 mg/kg) in wild-type mice, and was absent in CB2 (-/-) mice. Conversely, the CB2 receptor agonist, O-1966 (1, 3, 5, 10, 20 mg/kg) given in combination with a subthreshold dose of nicotine (0.1 mg/kg) elicited a place preference. In contrast, O-1966 (20 mg/kg) blocked cocaine (10 mg/kg)-induced CPP in wild type mice, while CB2 (-/-) mice showed unaltered cocaine CPP. CB2 (+/+) and (-/-) nicotine-dependent mice showed almost identical precipitated withdrawal responses and deletion of CB2 receptor did not alter acute somatic effects of nicotine. CONCLUSIONS: Collectively, these results indicate that CB2 receptors are required for nicotine-induced CPP in the mouse, while it is not involved in nicotine withdrawal or acute effects of nicotine. Moreover, these results suggest that CB2 receptors play opposing roles in nicotine- and cocaine-induced CPP.


Assuntos
Cocaína/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Nicotina/farmacologia , Receptor CB2 de Canabinoide/metabolismo , Animais , Anisóis/administração & dosagem , Anisóis/farmacologia , Comportamento Animal/efeitos dos fármacos , Cicloexanóis , Relação Dose-Resposta a Droga , Feminino , Masculino , Mecamilamina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nicotina/administração & dosagem , Receptor CB2 de Canabinoide/genética , Recompensa , Síndrome de Abstinência a Substâncias/metabolismo , Tabagismo/metabolismo
18.
Neuropharmacology ; 70: 228-35, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23416040

RESUMO

The 15q25 gene cluster contains genes that code for the α5, α3, and ß4 nicotinic acetylcholine receptor (nAChRs) subunits, and in human genetic studies, has shown the most robust association with smoking behavior and nicotine dependence to date. The limited available animal studies implicate a role for the α5 and ß4 nAChR subunits in nicotine dependence and withdrawal; however studies focusing on the behavioral role of the α3ß4* nAChR receptor subtype in nicotine dependence are lacking. Because of the apparent role of the α3ß4* nAChR subtype in nicotine dependence, the goal of the current study was to better evaluate the involvement of this subtype in nicotine mediated behavioral responses. Using the selective α3ß4* nAChR antagonist, α-conotoxin AuIB, we assessed the role of α3ß4* nAChRs in acute nicotine, nicotine reward, and physical and affective nicotine withdrawal. Because α5 has also been implicated in nicotine dependence behaviors in mice and can form functional receptors with α3ß4*, we also evaluated the role of the α3ß4α5* nAChR subtype in nicotine reward and somatic nicotine withdrawal signs by blocking the α3ß4* nAChR subtype in α5 nAChR knockout mice with AuIB. AuIB had no significant effect on acute nicotine behaviors, but dose-dependently attenuated nicotine reward and physical withdrawal signs, with no significant effect in affective withdrawal measures. Interestingly, AuIB also attenuated nicotine reward and somatic signs in α5 nAChR knockout mice. This study shows that α3ß4* nAChRs mediate nicotine reward and physical nicotine withdrawal, but not acute nicotine behaviors or affective nicotine withdrawal signs in mice. The α5 subunit is not required in the receptor assembly to mediate these effects. Our findings suggest an important role for the α3ß4* nAChR subtype in nicotine reward and physical aspects of the nicotine withdrawal syndrome.


Assuntos
Nicotina/farmacologia , Receptores Nicotínicos/fisiologia , Recompensa , Síndrome de Abstinência a Substâncias/fisiopatologia , Animais , Conotoxinas/farmacologia , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Knockout , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Receptores Nicotínicos/genética , Síndrome de Abstinência a Substâncias/psicologia
19.
Life Sci ; 92(8-9): 458-62, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-22705310

RESUMO

The endogenous cannabinoid anandamide (AEA) exerts the majority of its effects at CB1 and CB2 receptors and is degraded by fatty acid amide hydrolase (FAAH). FAAH KO mice and animals treated with FAAH inhibitors are impaired in their ability to hydrolyze AEA and other non-cannabinoid lipid signaling molecules, such as oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). AEA and these other substrates activate non-cannabinoid receptor systems, including TRPV1 and PPAR-α receptors. In this mini review, we describe the functional consequences of FAAH inhibition on nicotine reward and dependence as well as the underlying endocannabinoid and non-cannabinoid receptor systems mediating these effects. Interestingly, FAAH inhibition seems to mediate nicotine dependence differently in mice and rats. Indeed, pharmacological and genetic FAAH disruption in mice enhances nicotine reward and withdrawal. However, in rats, pharmacological blockade of FAAH significantly inhibits nicotine reward and has no effect in nicotine withdrawal. Studies suggest that non-cannabinoid mechanisms may play a role in these species differences.


Assuntos
Amidoidrolases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Tabagismo/tratamento farmacológico , Animais , Endocanabinoides/fisiologia , Humanos , Camundongos , Ratos , Recompensa , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Síndrome de Abstinência a Substâncias/fisiopatologia , Tabagismo/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...